Broad and Differential Animal ACE2 Receptor Usage by SARS-Cov-2

Total Page:16

File Type:pdf, Size:1020Kb

Broad and Differential Animal ACE2 Receptor Usage by SARS-Cov-2 bioRxiv preprint doi: https://doi.org/10.1101/2020.04.19.048710; this version posted April 19, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 Broad and differential animal ACE2 receptor usage by SARS-CoV-2 2 3 Xuesen Zhao1, 2#*, Danying Chen1, 2#, Robert Szabla3, Mei Zheng1, 2, Guoli Li1, 2, 4 Pengcheng Du1, 2, Shuangli Zheng1, 2, Xinglin Li1, 2, Chuan Song1, 2, Rui Li1, 2, Ju-Tao 5 Guo4, Murray Junop3, Hui Zeng1, 2*, Hanxin Lin5* 6 1Institute of Infectious disease, Beijing Ditan Hospital, Capital Medical University, 7 Beijing 100015, China. 8 2Beijing Key Laboratory of Emerging Infectious Disease, Beijing 100015, China. 9 3Department of Biochemistry, Western University, 1151 Richmond Street, London, 10 Ontario, Canada. 11 4Baruch S. Blumberg Institute, Hepatitis B Foundation, 3805 Old Easton Road, 12 Doylestown, PA 18902. USA. 13 5Department of Pathology and Laboratory Medicine, Western University, 1151 14 Richmond Street, London, Ontario, Canada. 15 16 Running Title: Broad animal ACE2 receptor usage by SARS-CoV-2 17 18 * Corresponding author’s e-mail address: 19 Xuesen Zhao, [email protected] 20 Hui Zeng, [email protected] 21 Hanxin Lin, [email protected] 22 # These authors are co-first authors. 23 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.04.19.048710; this version posted April 19, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 24 ABSTRACT 25 The COVID-19 pandemic has caused an unprecedented global public health and 26 economy crisis. The origin and emergence of its causal agent, SARS-CoV-2, in the 27 human population remains mysterious, although bat and pangolin were proposed to be 28 the natural reservoirs. Strikingly, comparing to the SARS-CoV-2-like CoVs identified in 29 bats and pangolins, SARS-CoV-2 harbors a polybasic furin cleavage site in its spike (S) 30 glycoprotein. SARS-CoV-2 uses human ACE2 as its receptor to infect cells. Receptor 31 recognition by the S protein is the major determinant of host range, tissue tropism, and 32 pathogenesis of coronaviruses. In an effort to search for the potential intermediate or 33 amplifying animal hosts of SARS-CoV-2, we examined receptor activity of ACE2 from 34 14 mammal species and found that ACE2 from multiple species can support the 35 infectious entry of lentiviral particles pseudotyped with the wild-type or furin cleavage 36 site deficient S protein of SARS-CoV-2. ACE2 of human/rhesus monkey and rat/mouse 37 exhibited the highest and lowest receptor activity, respectively. Among the remaining 38 species, ACE2 from rabbit and pangolin strongly bound to the S1 subunit of 39 SARS-CoV-2 S protein and efficiently supported the pseudotyped virus infection. These 40 findings have important implications for understanding potential natural reservoirs, 41 zoonotic transmission, human-to-animal transmission, and use of animal models. 42 43 44 45 Key words: SARS-CoV-2; animal ACE2; receptor; entry; furin cleavage; animal hosts 46 2 bioRxiv preprint doi: https://doi.org/10.1101/2020.04.19.048710; this version posted April 19, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 47 Introduction 48 49 Coronavirus disease 2019 (COVID-19) was first identified in Dec. 2019 in the city 50 of Wuhan, China 1, and has since spread worldwide, causing 2.3 million infected and 51 around 160,000 fatalities as of April 18th, 2020 (https://coronavirus.jhu.edu/map.html). 52 These numbers are still growing rapidly. The global COVID-19 pandemic has caused an 53 unprecedented public health and economy crisis. 54 COVID-19 is caused by a novel coronavirus, Severe Acute Respiratory Syndrome 55 Coronavirus 2 (SARS-CoV-2; initially named as 2019-nCoV) 2,3. The origin of 56 SARS-CoV-2 and its emergence in the human population remain mysterious. Many of 57 the early cases were linked to the Huanan seafood and wild animal market in Wuhan 58 city, raising the possibility of zoonotic origin 4. Sequencing analyses showed that the 59 genome of SARS-CoV-2 shares 79.5%, 89.1%, 93.3%, and 96.2% nucleotide sequence 60 identity with that of human SARS-CoV, bat coronavirus (CoV) ZC45, bat CoV 61 RmYN02, and bat CoV RaTG13, respectively, suggesting that SARS-CoV-2 probably 62 has bat origins 2,3,5. This finding is not surprising as bats are notorious for serving as the 63 natural reservoir for many emerging zoonotic viral pathogens, including two other 64 deadly human coronaviruses, SARS-CoV and Middle East respiratory syndrome 65 coronavirus (MERS-CoV), which caused global outbreak during 2002-2003 and 66 2012-2015, respectively 6,7. 67 Although SARS-CoV-2 may have originated from bats, bat CoVs are unlikely to 68 jump directly to humans due to the general ecological separation. Other mammal 69 species may have been served as intermediate or amplifying hosts where the progenitor 3 bioRxiv preprint doi: https://doi.org/10.1101/2020.04.19.048710; this version posted April 19, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 70 virus acquires critical mutations for efficient zoonotic transmission to human. This has 71 been seen in the emergence of SARS-CoV and MERS-CoV where palm civet and 72 dromedary camel act as the respective intermediate host 7. The Huanan seafood and 73 wild animal market in Wuhan city would otherwise be a unique place to trace any 74 potential animal source; however, soon after the disease outbreak, the market was 75 closed and all the wild animals were cleared, making this task very challenging or even 76 impossible. As an alternative, wide screening of wild animals becomes imperative. 77 Several recent studies identified multiple SARS-COV-2-like CoVs (SL-CoVs) from 78 smuggled Malayan pangolins in China. These pangolin CoVs (PCoV) form two 79 phylogenetic lineages, PCoV-GX and PCoV-GD 8-11. In particular, lineage PCoV-GD 80 was found to carry a nearly identical receptor-binding motif (RBM) in the spike (S) 81 protein to that of SARS-CoV-2 (Fig.1). However, the genome of these pangolin 82 SL-CoVs share only 85.5%-92.4% nucleotide identities with that of SARS-CoV-2. This 83 is in contrast to SARS-CoV and MERS-CoV where CoVs isolated form the 84 intermediate host palm civet and dromedary camel share 99.6% and 99.9% % genome 85 sequence identities with their human counterpart, respectively 12,13. Therefore, pangolins 86 tested in these studies are not the direct intermediate host for SARS-CoV-2. Whether or 87 not SARS-CoV-2 came from other pangolins or other wild animal species remains to be 88 determined. 89 Receptor recognition by the viral S protein is the major determinant of host range, 90 cell, tissue tropism, and pathogenesis of coronaviruses 14. The S protein of 91 SARS-CoV-2 is a type I membrane glycoprotein, which can be cleaved to S1 and S2 92 subunit during biogenesis at the polybasic furin cleavage site (RRAR) (Fig.1) 15-17. 4 bioRxiv preprint doi: https://doi.org/10.1101/2020.04.19.048710; this version posted April 19, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 93 Previous studies have shown that furin cleavage is not essential for coronavirus-cell 94 membrane fusion, but enhances cell-to-cell fusion 18-22, increases the fitness of sequence 95 variant within the quasispecies population of bovine CoV 23, and convert avirulent avian 96 influenza virus isolate to a highly pathogenic isolate 24. Interestingly, this cleavage site 97 is not present in the S protein of SARS-CoV, bat SL-CoVs or pangolin SL-CoVs 98 identified so far 5,15. During cell entry, S1 binds to the cellular receptor, subsequently 99 triggering a cascade of events leading to S2-mediated membrane fusion between host 100 cells and coronavirus particles 25. S1 protein contains an independently folded domain 101 called the receptor binding domain (RBD), which harbors an RBM that is primarily 102 involved in contact with receptor (Fig. 1). Human ACE2 (hACE2) has been identified as 103 the cellular receptor for both SARS-CoV-2 3,15,17,26 and SARS-CoV 27. In addition to 104 hACE2, ACE2 from horseshoe bat (Rhinolophus alcyone) was found to support cell 105 entry of SARS-CoV-2 S-mediated VSV-based pseudotyped virus 15. By using infectious 106 virus it has also been shown that ACE2 from Chinese horseshoe bat (Rhinolophus 107 sinicus), civet and swine, but not mouse, could serve as functional receptors 3. However, 108 in this infection system, the entry step was coupled with other steps during virus life 109 cycle, i.e.
Recommended publications
  • Multiple Recombination Events and Strong Purifying Selection at the Origin of SARS-Cov-2 Spike Glycoprotein Increased Correlated Dynamic Movements
    International Journal of Molecular Sciences Article Multiple Recombination Events and Strong Purifying Selection at the Origin of SARS-CoV-2 Spike Glycoprotein Increased Correlated Dynamic Movements Massimiliano S. Tagliamonte 1,2,† , Nabil Abid 3,4,†, Stefano Borocci 5,6 , Elisa Sangiovanni 5, David A. Ostrov 2 , Sergei L. Kosakovsky Pond 7, Marco Salemi 1,2,*, Giovanni Chillemi 5,8,*,‡ and Carla Mavian 1,2,*,‡ 1 Emerging Pathogen Institute, University of Florida, Gainesville, FL 32608, USA; mstagliamonte@ufl.edu 2 Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; [email protected]fl.edu 3 Laboratory of Transmissible Diseases and Biological Active Substances LR99ES27, Faculty of Pharmacy, University of Monastir, Rue Ibn Sina, 5000 Monastir, Tunisia; [email protected] 4 Department of Biotechnology, High Institute of Biotechnology of Sidi Thabet, University of Manouba, BP-66, 2020 Ariana-Tunis, Tunisia 5 Department for Innovation in Biological, Agro-food and Forest Systems (DIBAF), University of Tuscia, via S. Camillo de Lellis s.n.c., 01100 Viterbo, Italy; [email protected] (S.B.); [email protected] (E.S.) 6 Institute for Biological Systems, National Research Council, Via Salaria, Km 29.500, 00015 Monterotondo, Rome, Italy 7 Department of Biology, Temple University, Philadelphia, PA 19122, USA; [email protected] 8 Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Via Giovanni Amendola, 122/O, 70126 Bari, Italy * Correspondence: [email protected]fl.edu (M.S.); [email protected] (G.C.); cmavian@ufl.edu (C.M.) † Authors contributed equally as first to this work. ‡ Authors contributed equally as last to this work.
    [Show full text]
  • Inverted Repeats in Coronavirus SARS-Cov-2 Genome and Implications in Evolution
    Inverted repeats in coronavirus SARS-CoV-2 genome and implications in evolution Changchuan Yin ID ∗ Department of Mathematics, Statistics, and Computer Science University of Illinois at Chicago Chicago, IL 60607 USA Stephen S.-T. Yau ID y Department of Mathematical Sciences Tsinghua University Beijing 100084 China Abstract The coronavirus disease (COVID-19) pandemic, caused by the coronavirus SARS- CoV-2, has caused 60 millions of infections and 1.38 millions of fatalities. Genomic analysis of SARS-CoV-2 can provide insights on drug design and vaccine develop- ment for controlling the pandemic. Inverted repeats in a genome greatly impact the stability of the genome structure and regulate gene expression. Inverted repeats involve cellular evolution and genetic diversity, genome arrangements, and diseases. Here, we investigate the inverted repeats in the coronavirus SARS-CoV-2 genome. We found that SARS-CoV-2 genome has an abundance of inverted repeats. The inverted repeats are mainly located in the gene of the Spike protein. This result suggests the Spike protein gene undergoes recombination events, therefore, is essential for fast evolution. Comparison of the inverted repeat signatures in human and bat coronaviruses suggest that SARS-CoV-2 is mostly related SARS-related coronavirus, SARSr-CoV/RaTG13. The study also reveals that the recent SARS- related coronavirus, SARSr-CoV/RmYN02, has a high amount of inverted repeats in the spike protein gene. Besides, this study demonstrates that the inverted repeat distribution in a genome can be considered as the genomic signature. This study highlights the significance of inverted repeats in the evolution of SARS-CoV-2 and presents the inverted repeats as the genomic signature in genome analysis.
    [Show full text]
  • Exploring the Natural Origins of SARS-Cov-2
    bioRxiv preprint doi: https://doi.org/10.1101/2021.01.22.427830; this version posted January 22, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Exploring the natural origins of SARS-CoV-2 1 1 2 3 1,* Spyros Lytras ,​ Joseph Hughes ,​ Wei Xia ,​ Xiaowei Jiang ,​ David L Robertson ​ ​ ​ ​ ​ 1 MRC-University​ of Glasgow Centre for Virus Research (CVR), Glasgow, UK. 2 National​ School of Agricultural Institution and Development, South China Agricultural University, Guangzhou, China. 3 Department​ of Biological Sciences, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China. * Correspondence:​ [email protected] Summary. The lack of an identifiable intermediate host species for the proximal animal ​ ancestor of SARS-CoV-2 and the distance (~1500 km) from Wuhan to Yunnan province, where the closest evolutionary related coronaviruses circulating in horseshoe bats have been identified, is fueling speculation on the natural origins of SARS-CoV-2. Here we analyse SARS-CoV-2’s related horseshoe bat and pangolin Sarbecoviruses and confirm ​ Rhinolophus affinis continues to be the likely reservoir species as its host range extends ​ across Central and Southern China. This would explain the bat Sarbecovirus recombinants ​ in the West and East China, trafficked pangolin infections and bat Sarbecovirus recombinants linked to Southern China. Recent ecological disturbances as a result of changes in meat consumption could then explain SARS-CoV-2 transmission to humans through direct or indirect contact with the reservoir wildlife, and subsequent emergence towards Hubei in Central China.
    [Show full text]
  • WHO-Convened Global Study of Origins of SARS-Cov-2: China Part
    WHO-convened Global Study of Origins of SARS-CoV-2: China Part Joint WHO-China Study 14 January-10 February 2021 Joint Report 1 LIST OF ABBREVIATIONS AND ACRONYMS ARI acute respiratory illness cDNA complementary DNA China CDC Chinese Center for Disease Control and Prevention CNCB China National Center for Bioinformation CoV coronavirus Ct values cycle threshold values DDBJ DNA Database of Japan EMBL-EBI European Molecular Biology Laboratory and European Bioinformatics Institute FAO Food and Agriculture Organization of the United Nations GISAID Global Initiative on Sharing Avian Influenza Database GOARN Global Outbreak Alert and Response Network Hong Kong SAR Hong Kong Special Administrative Region Huanan market Huanan Seafood Wholesale Market IHR International Health Regulations (2005) ILI influenza-like illness INSD International Nucleotide Sequence Database MERS Middle East respiratory syndrome MRCA most recent common ancestor NAT nucleic acid testing NCBI National Center for Biotechnology Information NMDC National Microbiology Data Center NNDRS National Notifiable Disease Reporting System OIE World Organisation for Animal Health (Office international des Epizooties) PCR polymerase chain reaction PHEIC public health emergency of international concern RT-PCR real-time polymerase chain reaction SARI severe acute respiratory illness SARS-CoV-2 Severe acute respiratory syndrome coronavirus 2 SARSr-CoV-2 Severe acute respiratory syndrome coronavirus 2-related virus tMRCA time to most recent common ancestor WHO World Health Organization WIV Wuhan Institute of Virology 2 Acknowledgements WHO gratefully acknowledges the work of the joint team, including Chinese and international scientists and WHO experts who worked on the technical sections of this report, and those who worked on studies to prepare data and information for the joint mission.
    [Show full text]
  • Science Journals
    RESEARCH CORONAVIRUS ticles showed enhanced heterologous binding and neutralization properties against hu- Mosaic nanoparticles elicit cross-reactive immune man and bat SARS-like betacoronaviruses (sarbecoviruses). responses to zoonotic coronaviruses in mice We used a study of sarbecovirus RBD re- ceptor usage and cell tropism (38) to guide Alexander A. Cohen1, Priyanthi N. P. Gnanapragasam1, Yu E. Lee1, Pauline R. Hoffman1, Susan Ou1, our choice of RBDs for co-display on mosaic Leesa M. Kakutani1, Jennifer R. Keeffe1, Hung-Jen Wu2, Mark Howarth2, Anthony P. West1, particles. From 29 RBDs that were classified Christopher O. Barnes1, Michel C. Nussenzweig3, Pamela J. Bjorkman1* into distinct clades (clades 1, 2, 1/2, and 3) (38), we identified diverse RBDs from SARS- Protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and SARS-related emergent CoV, WIV1, and SHC014 (clade 1); SARS-CoV-2 zoonotic coronaviruses is urgently needed. We made homotypic nanoparticles displaying the receptor binding (clade 1/2); Rs4081, Yunnan 2011 (Yun11), and domain (RBD) of SARS-CoV-2 or co-displaying SARS-CoV-2 RBD along with RBDs from animal betacoronaviruses Rf1 (clade 2); and BM-4831 (clade 3). Of these, that represent threats to humans (mosaic nanoparticles with four to eight distinct RBDs). Mice immunized SARS-CoV-2 and SARS-CoV are human coro- with RBD nanoparticles, but not soluble antigen, elicited cross-reactive binding and neutralization responses. naviruses and the rest are bat viruses originat- Mosaic RBD nanoparticles elicited antibodies with superior cross-reactive recognition of heterologous RBDs ing in China or Bulgaria (BM-4831). We also relative to sera from immunizations with homotypic SARS-CoV-2–RBD nanoparticles or COVID-19 convalescent included RBDs from the GX pangolin clade 1/2 human plasmas.
    [Show full text]
  • The SARS-Cov-2 Arginine Dimers
    The SARS-CoV-2 arginine dimers Antonio R. Romeu ( [email protected] ) University Rovira i Virgili Enric Ollé University Rovira i Virgili Short Report Keywords: SARS-CoV-2, Sarbecovirus, Arginine dimer, Arginine codons usage, Bioinformatics Posted Date: August 2nd, 2021 DOI: https://doi.org/10.21203/rs.3.rs-770380/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License The SARS-CoV-2 arginine dimers Antonio R. Romeu1 and Enric Ollé2 1: Chemist. Professor of Biochemistry and Molecular Biology. University Rovira i Virgili. Tarragona. Spain. Corresponding author. Email: [email protected] 2: Veterinarian, Biochemist. Associate Professor of the Department of Biochemistry and Biotechnology. University Rovira i Virgili. Tarragona. Spain. Email: [email protected] Abstract Arginine is present, even as a dimer, in the viral polybasic furin cleavage sites, including that of SARS-CoV-2 in its protein S, whose acquisition is one of its characteristics that distinguishes it from the rest of the sarbecoviruses. The CGGCGG sequence encodes the SARS-CoV-2 furin site RR dimer. The aim of this work is to report the other SARS-CoV-2 arginine pairs, with particular emphasis in their codon usage. Here we show the presence of RR dimers in the orf1ab related non structural proteins nsp3, nsp4, nsp6, nsp13 and nsp14A2. Also, with a higher proportion in the structural roteinp nucleocapsid. All these RR dimers were strictly conserved in the sarbecovirus strains closest to SARS-CoV-2, and none of them was encoded by the CGGCGG sequence. Key words SARS-CoV-2, Sarbecovirus, Arginine dimer, Arginine codons usage, Bioinformatics Introduction Arginine (R) is a polar and non-hydrophobic amino acid, with a positive charged guanidine group, a physiological pH, linked a 3-hydrocarbon aliphatic chain.
    [Show full text]
  • To Stop the Next Pandemic, We Need to Unravel the Origins of COVID-19 David A
    OPINION OPINION To stop the next pandemic, we need to unravel the origins of COVID-19 David A. Relmana,b,c,d,1 We find ourselves ten months into one of the most to have been collected from bats in 2013 and 2019, catastrophic global health events of our lifetime and, respectively, in Yunnan Province, China (1). COVID-19 disturbingly, we still do not know how it began. What’s was first reported in December 2019 more than 1,000 even more troubling is that despite the critical impor- miles away in Wuhan City, Hubei Province, China. tance of this question, efforts to investigate the origins Beyond these facts, the “origin story” is missing many of the severe acute respiratory syndrome coronavirus key details, including a plausible and suitably detailed 2 (SARS-CoV-2) virus and of the associated disease, recent evolutionary history of the virus, the identity coronavirus disease 2019 (COVID-19), have become and provenance of its most recent ancestors, and sur- mired in politics, poorly supported assumptions and prisingly, the place, time, and mechanism of transmis- assertions, and incomplete information. sion of the first human infection. Even though a SARS-CoV-2 is a betacoronavirus whose apparent definitive answer may not be forthcoming, and even closest relatives, RaTG13 and RmYN02, are reported though an objective analysis requires addressing To avoid or mitigate the dire consequences of this and future pandemics (here, people in PPE bury a victim in Delhi, India in June), unraveling the origins of SARS-CoV-2 and COVID-19 will be essential—even though a definitive answer may be elusive, and an objective analysis means broaching some uncomfortable possibilities.
    [Show full text]
  • Unique Features Concerning the Proximal Origin of SARS-Cov-2
    Unique features concerning the proximal origin of SARS-CoV-2 Murat Seyran1,2, Damiano Pizzol3, Parise Adadi4, Tarek Mohamed Abd El-Aziz5,6, Sk. Sarif Hassan7, Antonio Soares5, Ramesh Kandimalla8,9, Kenneth Lundstrom10, Murtaza Tambuwala11, Alaa A. A. Aljabali 12, Amos Lal13, Gajendra Kumar Azad14, Pabitra Pal Choudhury15, Vladimir N. Uversky16, Samendra P. Sherchan17, Bruce D. Uhal18, Nima Rezaei19,20,*, Adam M. Brufsky21,* 1Doctoral studies in natural and technical sciences (SPL 44), University of Vienna, Austria. 2Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Austria. [email protected], [email protected] 3Italian Agency for Development Cooperation - Khartoum, Sudan Street 33, Al Amarat, Sudan. [email protected] 4Department of Food Science, University of Otago, Dunedin 9054, New Zealand. [email protected], [email protected] 5Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229-3900, USA. [email protected], [email protected] 6Zoology Department, Faculty of Science, Minia University, El-Minia 61519, Egypt. 7Department of Mathematics, Pingla Thana Mahavidyalaya, Maligram, Paschim Medinipur, 721140, West Bengal, India. [email protected] 8CSIR-Indian Institute of Chemical Technology Uppal Road, Tarnaka, Hyderabad-500007, Telangana State, India. 9Kakatiya Medical College/MGM-Hospital, DME/TSPSC, Hyderabad, Warangal, Telangana State- 506007, India. [email protected], [email protected] 10PanTherapeutics, Rte de Lavaux 49, CH1095 Lutry, Switzerland. [email protected] 11School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, Northern Ireland, UK. [email protected] 12Department of Pharmaceutical Sciences, Yarmouk University—Faculty of Pharmacy, Irbid 566, Jordan 1 13Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota, USA.
    [Show full text]
  • The SARS-Cov-2 Genome: Variation, Implication and Application
    26 AUGUST 2020 The SARS-CoV-2 genome: variation, implication and application This rapid review describes the severe acute respiratory syndrome coronavirus-2 (SARS- CoV-2) genome, its relationship to other coronaviruses, the variation that has occurred since SARS-CoV-2 emerged in Wuhan in late 2019, the implications of these changes and how knowledge of these changes may be utilised. This pre-print from the Royal Society is provided to assist in the understanding of COVID-19. Executive summary • SARS-CoV-2 emerged in late 2019 in Wuhan, China. The • Whole genome sequencing is a valuable addition to test, genome of many separate virus isolates from early in the track and trace, and is encouraged. For instance it: Wuhan outbreak are very closely related showing the virus – has revealed >1350 separate introductions of SARS- emerged recently in humans. CoV-2 into the UK from mid-February to mid-March • The SARS-CoV-2 genome is sufficiently different to all 2020 arising very largely from Spain, France and known coronaviruses to refute the assertion that the Italy, and not from China. COVID-19 pandemic arose by deliberate or accidental – can be used to follow transmission within specific release of a known virus and make it highly improbable communities such as hospitals, schools or factories, that the virus arose by artificial construction in a laboratory. and combined with epidemiological data enables • SARS-CoV-2 is most closely related to bat coronaviruses routes of transmission to be identified and barriers from China, but even the closest of these viruses are too to transmission implemented.
    [Show full text]
  • Genetic Spectrum and Distinct Evolution Patterns of SARS-Cov-2
    medRxiv preprint doi: https://doi.org/10.1101/2020.06.16.20132902; this version posted August 5, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY-NC-ND 4.0 International license . Genetic spectrum and distinct evolution patterns of SARS-CoV-2 Sheng Liu1,2 ¶, Jikui Shen3 ¶, Shuyi Fang4, Kailing Li4, Juli Liu5, Lei Yang5, Chang-Deng Hu6,7, Jun Wan1,2,4,8 * 1. Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA 2. Collaborative Core for Cancer Bioinformatics (C3B) shared by Indiana University Simon Comprehensive Cancer Center and Purdue University Center for Cancer Research, Indiana USA 3. The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA 4. Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indiana University – Purdue University Indianapolis, Indianapolis, Indiana, USA 5. Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana USA 6. Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA 7. Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana USA 8. The Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA ¶ These authors have equal contributions. * To whom correspondence should be addressed. Tel: 1-317-2786445; Fax: 1-317-278-9217; Email: [email protected] Keywords SARS-CoV-2, single nucleotide variant (SNV), clustering, co-infection, evolution. Abstract Four signature groups of frequently occurred single-nucleotide variants (SNVs) were identified in over twenty-eight thousand high-quality and high-coverage SARS-CoV-2 complete genome sequences, representing different viral strains.
    [Show full text]
  • WHO-Convened Global Study of the Origins of SARS-Cov-2: Terms of References for the China Part
    Terms of references – FINAL DRAFT WHO-convened Global Study of the Origins of SARS-CoV-2: Terms of References for the China Part 31 July 2020 Contents Background ............................................................................................................................................ 2 Approach ................................................................................................................................................ 2 Current knowledge supporting origin tracing work ............................................................................... 3 Phylogenetic work.............................................................................................................................. 3 Animal susceptibility and animal surveys .......................................................................................... 4 Persistence of the virus on food and surfaces ................................................................................... 4 The early cluster in Wuhan ................................................................................................................ 5 Implementation plan ............................................................................................................................. 6 Phase 1: Short term studies ............................................................................................................... 6 a) Descriptive Epidemiological Study ........................................................................................
    [Show full text]
  • Evidence for SARS-Cov-2 Related Coronaviruses Circulating in Bats and Pangolins in Southeast Asia
    ARTICLE https://doi.org/10.1038/s41467-021-21240-1 OPEN Evidence for SARS-CoV-2 related coronaviruses circulating in bats and pangolins in Southeast Asia Supaporn Wacharapluesadee1,10, Chee Wah Tan 2,10, Patarapol Maneeorn3,10, Prateep Duengkae4, Feng Zhu 2, Yutthana Joyjinda1, Thongchai Kaewpom1, Wan Ni Chia2, Weenassarin Ampoot1, Beng Lee Lim2, Kanthita Worachotsueptrakun1, Vivian Chih-Wei Chen 2, Nutthinee Sirichan4, Chanida Ruchisrisarod1, Apaporn Rodpan1, Kirana Noradechanon3, Thanawadee Phaichana3, Niran Jantarat3, Boonchu Thongnumchaima3, Changchun Tu 5,6, Gary Crameri7, Martha M. Stokes8, ✉ ✉ Thiravat Hemachudha1,11 & Lin-Fa Wang 2,9,11 1234567890():,; Among the many questions unanswered for the COVID-19 pandemic are the origin of SARS- CoV-2 and the potential role of intermediate animal host(s) in the early animal-to-human transmission. The discovery of RaTG13 bat coronavirus in China suggested a high probability of a bat origin. Here we report molecular and serological evidence of SARS-CoV-2 related coronaviruses (SC2r-CoVs) actively circulating in bats in Southeast Asia. Whole genome sequences were obtained from five independent bats (Rhinolophus acuminatus) in a Thai cave yielding a single isolate (named RacCS203) which is most related to the RmYN02 isolate found in Rhinolophus malayanus in Yunnan, China. SARS-CoV-2 neutralizing antibodies were also detected in bats of the same colony and in a pangolin at a wildlife checkpoint in Southern Thailand. Antisera raised against the receptor binding domain (RBD) of RmYN02 was able to cross-neutralize SARS-CoV-2 despite the fact that the RBD of RacCS203 or RmYN02 failed to bind ACE2. Although the origin of the virus remains unresolved, our study extended the geographic distribution of genetically diverse SC2r-CoVs from Japan and China to Thailand over a 4800-km range.
    [Show full text]